1
|
Lewis CM, Griffith TN. Ion channels of cold transduction and transmission. J Gen Physiol 2024; 156:e202313529. [PMID: 39051992 PMCID: PMC11273221 DOI: 10.1085/jgp.202313529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/04/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Thermosensation requires the activation of a unique collection of ion channels and receptors that work in concert to transmit thermal information. It is widely accepted that transient receptor potential melastatin 8 (TRPM8) activation is required for normal cold sensing; however, recent studies have illuminated major roles for other ion channels in this important somatic sensation. In addition to TRPM8, other TRP channels have been reported to contribute to cold transduction mechanisms in diverse sensory neuron populations, with both leak- and voltage-gated channels being identified for their role in the transmission of cold signals. Whether the same channels that contribute to physiological cold sensing also mediate noxious cold signaling remains unclear; however, recent work has found a conserved role for the kainite receptor, GluK2, in noxious cold sensing across species. Additionally, cold-sensing neurons likely engage in functional crosstalk with nociceptors to give rise to cold pain. This Review will provide an update on our understanding of the relationship between various ion channels in the transduction and transmission of cold and highlight areas where further investigation is required.
Collapse
Affiliation(s)
- Cheyanne M Lewis
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Theanne N Griffith
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
2
|
Uchida K. TRPM3, TRPM4, and TRPM5 as thermo-sensitive channels. J Physiol Sci 2024; 74:43. [PMID: 39294615 PMCID: PMC11409758 DOI: 10.1186/s12576-024-00937-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 09/01/2024] [Indexed: 09/20/2024]
Abstract
Temperature detection is essential for the survival and perpetuation of any species. Thermoreceptors in the skin sense body temperature as well as the temperatures of ambient air and objects. Since Dr. David Julius and his colleagues discovered that TRPV1 is expressed in small-diameter primary sensory neurons, and activated by temperatures above 42 °C, 11 of thermo-sensitive TRP channels have been identified. TRPM3 expressed in sensory neurons acts as a sensor for noxious heat. TRPM4 and TRPM5 are Ca2⁺-activated monovalent cation channels, and their activity is drastically potentiated by temperature increase. This review aims to summarize the expression patterns, electrophysiological properties, and physiological roles of TRPM3, TRPM4, and TRPM5 associated with thermosensation.
Collapse
Affiliation(s)
- Kunitoshi Uchida
- Laboratory of Functional Physiology, Department of Environmental and Life Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Yada 52-1, Suruga-Ku, Shizuoka, Shizuoka, 422-8526, Japan.
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan.
| |
Collapse
|
3
|
Arcas JM, Oudaha K, González A, Fernández-Trillo J, Peralta FA, Castro-Marsal J, Poyraz S, Taberner F, Sala S, de la Peña E, Gomis A, Viana F. The ion channel TRPM8 is a direct target of the immunosuppressant rapamycin in primary sensory neurons. Br J Pharmacol 2024; 181:3192-3214. [PMID: 38741464 DOI: 10.1111/bph.16402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND AND PURPOSE The mechanistic target of rapamycin (mTOR) signalling pathway is a key regulator of cell growth and metabolism. Its deregulation is implicated in several diseases. The macrolide rapamycin, a specific inhibitor of mTOR, has immunosuppressive, anti-inflammatory and antiproliferative properties. Recently, we identified tacrolimus, another macrolide immunosuppressant, as a novel activator of TRPM8 ion channels, involved in cold temperature sensing, thermoregulation, tearing and cold pain. We hypothesized that rapamycin may also have agonist activity on TRPM8 channels. EXPERIMENTAL APPROACH Using calcium imaging and electrophysiology in transfected HEK293 cells and wildtype or Trpm8 KO mouse DRG neurons, we characterized rapamycin's effects on TRPM8 channels. We also examined the effects of rapamycin on tearing in mice. KEY RESULTS Micromolar concentrations of rapamycin activated rat and mouse TRPM8 channels directly and potentiated cold-evoked responses, effects also observed in human TRPM8 channels. In cultured mouse DRG neurons, rapamycin increased intracellular calcium levels almost exclusively in cold-sensitive neurons. Responses were markedly decreased in Trpm8 KO mice or by TRPM8 channel antagonists. Cutaneous cold thermoreceptor endings were also activated by rapamycin. Topical application of rapamycin to the eye surface evokes tearing in mice by a TRPM8-dependent mechanism. CONCLUSION AND IMPLICATIONS These results identify TRPM8 cationic channels in sensory neurons as novel molecular targets of the immunosuppressant rapamycin. These findings may help explain some of its therapeutic effects after topical application to the skin and the eye surface. Moreover, rapamycin could be used as an experimental tool in the clinic to explore cold thermoreceptors.
Collapse
Affiliation(s)
- José Miguel Arcas
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Khalid Oudaha
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Alejandro González
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Jorge Fernández-Trillo
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | | | - Júlia Castro-Marsal
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Seyma Poyraz
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Francisco Taberner
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Salvador Sala
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Elvira de la Peña
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Ana Gomis
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Félix Viana
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| |
Collapse
|
4
|
Luu DD, Ramesh N, Kazan IC, Shah KH, Lahiri G, Mana MD, Ozkan SB, Van Horn WD. Evidence that the cold- and menthol-sensing functions of the human TRPM8 channel evolved separately. SCIENCE ADVANCES 2024; 10:eadm9228. [PMID: 38905339 PMCID: PMC11192081 DOI: 10.1126/sciadv.adm9228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/16/2024] [Indexed: 06/23/2024]
Abstract
Transient receptor potential melastatin 8 (TRPM8) is a temperature- and menthol-sensitive ion channel that contributes to diverse physiological roles, including cold sensing and pain perception. Clinical trials targeting TRPM8 have faced repeated setbacks predominantly due to the knowledge gap in unraveling the molecular underpinnings governing polymodal activation. A better understanding of the molecular foundations between the TRPM8 activation modes may aid the development of mode-specific, thermal-neutral therapies. Ancestral sequence reconstruction was used to explore the origins of TRPM8 activation modes. By resurrecting key TRPM8 nodes along the human evolutionary trajectory, we gained valuable insights into the trafficking, stability, and function of these ancestral forms. Notably, this approach unveiled the differential emergence of cold and menthol sensitivity over evolutionary time, providing a fresh perspective on complex polymodal behavior. These studies provide a paradigm for understanding polymodal behavior in TRPM8 and other proteins with the potential to enhance our understanding of sensory receptor biology and pave the way for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Dustin D. Luu
- School of Molecular Sciences and The Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Nikhil Ramesh
- Department of Physics and Center for Biological Physics, Arizona State University, Tempe, AZ, USA
| | - I. Can Kazan
- Department of Physics and Center for Biological Physics, Arizona State University, Tempe, AZ, USA
| | - Karan H. Shah
- School of Molecular Sciences and The Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Gourab Lahiri
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Miyeko D. Mana
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - S. Banu Ozkan
- Department of Physics and Center for Biological Physics, Arizona State University, Tempe, AZ, USA
| | - Wade D. Van Horn
- School of Molecular Sciences and The Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
5
|
Kiper AK, Wegner S, Kadala A, Rinné S, Schütte S, Winter Z, Bertoune MAR, Touska F, Matschke V, Wrobel E, Streit AK, Lang F, Schmidt C, Schulze-Bahr E, Schäfer MKH, Voelkl J, Seebohm G, Zimmermann K, Decher N. KCNQ1 is an essential mediator of the sex-dependent perception of moderate cold temperatures. Proc Natl Acad Sci U S A 2024; 121:e2322475121. [PMID: 38857404 PMCID: PMC11194602 DOI: 10.1073/pnas.2322475121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/25/2024] [Indexed: 06/12/2024] Open
Abstract
Low temperatures and cooling agents like menthol induce cold sensation by activating the peripheral cold receptors TRPM8 and TRPA1, cation channels belonging to the TRP channel family, while the reduction of potassium currents provides an additional and/or synergistic mechanism of cold sensation. Despite extensive studies over the past decades to identify the molecular receptors that mediate thermosensation, cold sensation is still not fully understood and many cold-sensitive peripheral neurons do not express the well-established cold sensor TRPM8. We found that the voltage-gated potassium channel KCNQ1 (Kv7.1), which is defective in cardiac LQT1 syndrome, is, in addition to its known function in the heart, a highly relevant and sex-specific sensor of moderately cold temperatures. We found that KCNQ1 is expressed in skin and dorsal root ganglion neurons, is sensitive to menthol and cooling agents, and is highly sensitive to moderately cold temperatures, in a temperature range at which TRPM8 is not thermosensitive. C-fiber recordings from KCNQ1-/- mice displayed altered action potential firing properties. Strikingly, only male KCNQ1-/- mice showed substantial deficits in cold avoidance at moderately cold temperatures, with a strength of the phenotype similar to that observed in TRPM8-/- animals. While sex-dependent differences in thermal sensitivity have been well documented in humans and mice, KCNQ1 is the first gene reported to play a role in sex-specific temperature sensation. Moreover, we propose that KCNQ1, together with TRPM8, is a key instrumentalist that orchestrates the range and intensity of cold sensation.
Collapse
Affiliation(s)
- Aytug K. Kiper
- Institute for Physiology and Pathophysiology, Department of Vegetative Physiology and Center for Mind, Brain and Behavior, Philipps-University Marburg, 35032Marburg, Germany
| | - Sven Wegner
- Institute for Physiology and Pathophysiology, Department of Vegetative Physiology and Center for Mind, Brain and Behavior, Philipps-University Marburg, 35032Marburg, Germany
| | - Aklesso Kadala
- Department of Anesthesiology, University of Erlangen-Nürnberg, 91054Erlangen, Germany
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Department of Vegetative Physiology and Center for Mind, Brain and Behavior, Philipps-University Marburg, 35032Marburg, Germany
| | - Sven Schütte
- Institute for Physiology and Pathophysiology, Department of Vegetative Physiology and Center for Mind, Brain and Behavior, Philipps-University Marburg, 35032Marburg, Germany
| | - Zoltán Winter
- Department of Anesthesiology, University of Erlangen-Nürnberg, 91054Erlangen, Germany
| | - Mirjam A. R. Bertoune
- Institute for Anatomy and Cell Biology, Department of Medicinal Cellbiology and Center for Mind, Brain and Behavior, Philipps-University Marburg, 35032Marburg, Germany
| | - Filip Touska
- Department of Anesthesiology, University of Erlangen-Nürnberg, 91054Erlangen, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, 44801Bochum, Germany
| | - Eva Wrobel
- Faculty of Chemistry and Biochemistry, Department of Receptor Biochemistry, Ruhr-University Bochum, 44780Bochum, Germany
| | - Anne-Kathrin Streit
- Institute for Physiology and Pathophysiology, Department of Vegetative Physiology and Center for Mind, Brain and Behavior, Philipps-University Marburg, 35032Marburg, Germany
| | - Florian Lang
- Institute for Physiology I, Department of Physiology I, Eberhard Karls University Tübingen, 72074Tübingen, Germany
| | - Constanze Schmidt
- Department of Cardiology, University Hospital Heidelberg, 69120Heidelberg, Germany
| | - Eric Schulze-Bahr
- Department for Genetics of Heart Diseases (IfG), University Hospital Münster, 48149Münster, Germany
| | - Martin K.-H. Schäfer
- Institute for Anatomy and Cell Biology, Department of Medicinal Cellbiology and Center for Mind, Brain and Behavior, Philipps-University Marburg, 35032Marburg, Germany
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Department of Physiology, Johannes Kepler University Linz, 4040Linz, Austria
| | - Guiscard Seebohm
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, 44801Bochum, Germany
- Department for Genetics of Heart Diseases (IfG), University Hospital Münster, 48149Münster, Germany
| | - Katharina Zimmermann
- Department of Anesthesiology, University of Erlangen-Nürnberg, 91054Erlangen, Germany
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Department of Vegetative Physiology and Center for Mind, Brain and Behavior, Philipps-University Marburg, 35032Marburg, Germany
| |
Collapse
|
6
|
Trif C, Banica AM, Manolache A, Anghel SA, Huţanu DE, Stratulat T, Badea R, Oprita G, Selescu T, Petrescu SM, Sisignano M, Offermanns S, Babes A, Tunaru S. Inhibition of TRPM8 function by prostacyclin receptor agonists requires coupling to Gq/11 proteins. Br J Pharmacol 2024; 181:1438-1451. [PMID: 38044577 DOI: 10.1111/bph.16295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND AND PURPOSE The TRPM8 ion channel is involved in innocuous cold sensing and has a potent anti-inflammatory action. Its activation by lower temperature or chemical agonists such as menthol and icilin induces analgesic effects, reversing hypersensitivity and reducing chronic pain. On the other hand, prostacyclin (PGI2) enhances pain and inflammation by activating the IP receptors. Due to the critical roles of TRPM8 and IP receptors in the regulation of inflammatory pain, and considering their overlapping expression pattern, we analysed the functional interaction between human TRPM8 and IP receptors. EXPERIMENTAL APPROACH We transiently expressed human TRPM8 channels and IP receptors in HEK293T cells and carried out intracellular calcium and cAMP measurements. Additionally, we cultured neurons from the dorsal root ganglia (DRGs) of mice and determined the increase in intracellular calcium triggered by the TRPM8 agonist, icilin, in the presence of the IP receptor agonist cicaprost, the IP receptor antagonist Cay10441, and the Gq/11 inhibitor YM254890. KEY RESULTS Activation of IP receptors by selective agonists (cicaprost, beraprost, and iloprost) inhibited TRPM8 channel function, independently of the Gs-cAMP pathway. The potent inhibition of TRPM8 channels by IP receptor agonists involved Gq/11 coupling. These effects were also observed in neurons isolated from murine DRGs. CONCLUSIONS AND IMPLICATIONS Our results demonstrate an unusual signalling pathway of IP receptors by coupling to Gq/11 proteins to inhibit TRPM8 channel function. This pathway may contribute to a better understanding of the role of TRPM8 channels and IP receptors in regulating pain and inflammation.
Collapse
Affiliation(s)
- Cosmin Trif
- Cell Signalling Research Group, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Alexandra-Maria Banica
- Cell Signalling Research Group, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Alexandra Manolache
- Department of Anatomy, Physiology, and Biophysics, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Sorina Andreea Anghel
- Cell Signalling Research Group, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Debora-Elena Huţanu
- Department of Anatomy, Physiology, and Biophysics, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Teodora Stratulat
- Cell Signalling Research Group, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
- Department of Anatomy, Physiology, and Biophysics, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Rodica Badea
- Cell Signalling Research Group, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - George Oprita
- Department of Anatomy, Physiology, and Biophysics, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Tudor Selescu
- Department of Anatomy, Physiology, and Biophysics, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Stefana M Petrescu
- Cell Signalling Research Group, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, Frankfurt am Main, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Alexandru Babes
- Department of Anatomy, Physiology, and Biophysics, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Sorin Tunaru
- Cell Signalling Research Group, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
- Prothanor Biotech S.R.L., Bucharest, Romania
| |
Collapse
|
7
|
Yang T, Yang Z, Xin W, Feng Y, Kong X, Wang Y, Li H, Wen L, Zhou G. Biomimicking TRPM8: A Conversely Temperature-Dependent Nonionic Retrorse Nanochannel for Ion Flow Control. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38679867 DOI: 10.1021/acsami.4c02990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Ion channels play a crucial role in the transmembrane transport and signal transmission of substances. In animals, transient receptor potential vanilloid 1 (TRPV1) and transient receptor potential melastatin 8 (TRPM8) serve as temperature-sensing units in sensory nerve endings. TRPV1 allows cells to sense heat, while TRPM8 enables them to detect cold, both serving to protect living organisms from harmful substances and environments. However, almost all studies on artificial nanochannels have mainly focused on TRPV1-like "forward nanochannels" thus far, which are incapable of "backward" responding to heat. So, we constructed an innovational TRPM8-inspired "retrorse nanochannel" through internal modification of poly(acrylamide-co-acrylonitrile) [P(AAm-co-AN)] with an upper critical solution temperature (UCST). Our results demonstrated that the internally modified nanochannels exhibited rapid, stable, and reversible heat-closing capability and converse temperature dependence within the typical temperature range of 25-40 °C. The biomimetic ion channel can effectively function as a facile, precise, and reversible thermal gate for controlling the transport of ions and substances. It also offers a promising microscopic technology for managing thermal effects on the substance, fluid, energy, and even signal delivery.
Collapse
Affiliation(s)
- Tao Yang
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology and Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, P. R. China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, P. R. China
| | - Zelin Yang
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology and Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, P. R. China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, P. R. China
| | - Weiwen Xin
- Key Laboratory of Bio-inspired Materials and Interfacial Science Technical, Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Yuchen Feng
- School of Future Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Xiangyu Kong
- Key Laboratory of Bio-inspired Materials and Interfacial Science Technical, Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Yao Wang
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology and Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, P. R. China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, P. R. China
| | - Hao Li
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology and Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, P. R. China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, P. R. China
| | - Liping Wen
- Key Laboratory of Bio-inspired Materials and Interfacial Science Technical, Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Guofu Zhou
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology and Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, P. R. China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, P. R. China
| |
Collapse
|
8
|
Liénard MA, Baez-Nieto D, Tsai CC, Valencia-Montoya WA, Werin B, Johanson U, Lassance JM, Pan JQ, Yu N, Pierce NE. TRPA5 encodes a thermosensitive ankyrin ion channel receptor in a triatomine insect. iScience 2024; 27:109541. [PMID: 38577108 PMCID: PMC10993193 DOI: 10.1016/j.isci.2024.109541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024] Open
Abstract
As ectotherms, insects need heat-sensitive receptors to monitor environmental temperatures and facilitate thermoregulation. We show that TRPA5, a class of ankyrin transient receptor potential (TRP) channels absent in dipteran genomes, may function as insect heat receptors. In the triatomine bug Rhodnius prolixus (order: Hemiptera), a vector of Chagas disease, the channel RpTRPA5B displays a uniquely high thermosensitivity, with biophysical determinants including a large channel activation enthalpy change (72 kcal/mol), a high temperature coefficient (Q10 = 25), and in vitro temperature-induced currents from 53°C to 68°C (T0.5 = 58.6°C), similar to noxious TRPV receptors in mammals. Monomeric and tetrameric ion channel structure predictions show reliable parallels with fruit fly dTRPA1, with structural uniqueness in ankyrin repeat domains, the channel selectivity filter, and potential TRP functional modulator regions. Overall, the finding of a member of TRPA5 as a temperature-activated receptor illustrates the diversity of insect molecular heat detectors.
Collapse
Affiliation(s)
- Marjorie A. Liénard
- Department of Biology, Lund University, 22362 Lund, Sweden
- Department of Organismic and Evolutionary Biology and Museum of Comparative Zoology, Harvard University, Cambridge, MA 02138, USA
- Broad Institute, Cambridge, MA 02142, USA
| | - David Baez-Nieto
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA 02142, USA
| | - Cheng-Chia Tsai
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, NY 10027, USA
| | - Wendy A. Valencia-Montoya
- Department of Organismic and Evolutionary Biology and Museum of Comparative Zoology, Harvard University, Cambridge, MA 02138, USA
| | - Balder Werin
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, 22362 Lund, Sweden
| | - Urban Johanson
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, 22362 Lund, Sweden
| | - Jean-Marc Lassance
- Department of Organismic and Evolutionary Biology and Museum of Comparative Zoology, Harvard University, Cambridge, MA 02138, USA
- Laboratory of Evolutionary Neuroethology, GIGA Institute, University of Liège, 4000 Liège, Belgium
| | - Jen Q. Pan
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA 02142, USA
| | - Nanfang Yu
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, NY 10027, USA
| | - Naomi E. Pierce
- Department of Organismic and Evolutionary Biology and Museum of Comparative Zoology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
9
|
Yeh F. Temperature gating in thermoTRPs may depend on temperature-dependent heat capacity differences. Temperature (Austin) 2024; 11:183-186. [PMID: 39193044 PMCID: PMC11346518 DOI: 10.1080/23328940.2024.2321066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024] Open
|
10
|
Amaya-Rodriguez CA, Carvajal-Zamorano K, Bustos D, Alegría-Arcos M, Castillo K. A journey from molecule to physiology and in silico tools for drug discovery targeting the transient receptor potential vanilloid type 1 (TRPV1) channel. Front Pharmacol 2024; 14:1251061. [PMID: 38328578 PMCID: PMC10847257 DOI: 10.3389/fphar.2023.1251061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/14/2023] [Indexed: 02/09/2024] Open
Abstract
The heat and capsaicin receptor TRPV1 channel is widely expressed in nerve terminals of dorsal root ganglia (DRGs) and trigeminal ganglia innervating the body and face, respectively, as well as in other tissues and organs including central nervous system. The TRPV1 channel is a versatile receptor that detects harmful heat, pain, and various internal and external ligands. Hence, it operates as a polymodal sensory channel. Many pathological conditions including neuroinflammation, cancer, psychiatric disorders, and pathological pain, are linked to the abnormal functioning of the TRPV1 in peripheral tissues. Intense biomedical research is underway to discover compounds that can modulate the channel and provide pain relief. The molecular mechanisms underlying temperature sensing remain largely unknown, although they are closely linked to pain transduction. Prolonged exposure to capsaicin generates analgesia, hence numerous capsaicin analogs have been developed to discover efficient analgesics for pain relief. The emergence of in silico tools offered significant techniques for molecular modeling and machine learning algorithms to indentify druggable sites in the channel and for repositioning of current drugs aimed at TRPV1. Here we recapitulate the physiological and pathophysiological functions of the TRPV1 channel, including structural models obtained through cryo-EM, pharmacological compounds tested on TRPV1, and the in silico tools for drug discovery and repositioning.
Collapse
Affiliation(s)
- Cesar A. Amaya-Rodriguez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Departamento de Fisiología y Comportamiento Animal, Facultad de Ciencias Naturales, Exactas y Tecnología, Universidad de Panamá, Ciudad de Panamá, Panamá
| | - Karina Carvajal-Zamorano
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado Universidad Católica del Maule, Talca, Chile
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | - Melissa Alegría-Arcos
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado Universidad Católica del Maule, Talca, Chile
| |
Collapse
|
11
|
Huang J, Korsunsky A, Yazdani M, Chen J. Targeting TRP channels: recent advances in structure, ligand binding, and molecular mechanisms. Front Mol Neurosci 2024; 16:1334370. [PMID: 38273937 PMCID: PMC10808746 DOI: 10.3389/fnmol.2023.1334370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Transient receptor potential (TRP) channels are a large and diverse family of transmembrane ion channels that are widely expressed, have important physiological roles, and are associated with many human diseases. These proteins are actively pursued as promising drug targets, benefitting greatly from advances in structural and mechanistic studies of TRP channels. At the same time, the complex, polymodal activation and regulation of TRP channels have presented formidable challenges. In this short review, we summarize recent progresses toward understanding the structural basis of TRP channel function, as well as potential ligand binding sites that could be targeted for therapeutics. A particular focus is on the current understanding of the molecular mechanisms of TRP channel activation and regulation, where many fundamental questions remain unanswered. We believe that a deeper understanding of the functional mechanisms of TRP channels will be critical and likely transformative toward developing successful therapeutic strategies targeting these exciting proteins. This endeavor will require concerted efforts from computation, structural biology, medicinal chemistry, electrophysiology, pharmacology, drug safety and clinical studies.
Collapse
Affiliation(s)
- Jian Huang
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Aron Korsunsky
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Mahdieh Yazdani
- Modeling and Informatics, Merck & Co., Inc., West Point, PA, United States
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
12
|
Uchida K. Temperature-Dependent Activation of Thermosensitive Transient Receptor Potential Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1461:47-59. [PMID: 39289273 DOI: 10.1007/978-981-97-4584-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Temperature detection is essential for the survival and perpetuation of any species. Thermoreceptors in the skin sense the body temperature and also the temperatures of the ambient air and the objects. In 1997, Dr. David Julius and his colleagues found that a receptor expressed in small-diameter primary sensory neurons was activated by capsaicin (the pungent chemical in hot pepper). This receptor was also activated by temperature above 42 °C. That was the first time that a thermal receptor in primary sensory neurons has been identified. This receptor is named transient receptor potential vanilloid 1 (TRPV1). Now, 11 thermosensitive TRP channels are known. In this chapter, we summarize the reports and analyze thermosensitive TRP channels in a variety of ways to clarify the activation mechanisms by which temperature changes are sensed.
Collapse
Affiliation(s)
- Kunitoshi Uchida
- Division of Environmental and Life Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan.
| |
Collapse
|
13
|
Zeng L, Li A, Qu K, Zhang J, Huang B, Lei C. TRPM4 gene variation associated with climatic conditions in Chinese cattle. Anim Biotechnol 2023; 34:3256-3260. [PMID: 35994677 DOI: 10.1080/10495398.2022.2112686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
The transient receptor potential (TRP) superfamily has been reported to play an important role in heat tolerance pathways. Based on the Bovine Genome Variation Database and Selective Signatures, a missense mutation (NC_037345.1: c.2237A > G: p. His746Arg) (rs209689836) was identified in the transient receptor potential cation channel subfamily M member 4 (TRPM4) gene, a member of the TRP family, corresponding to heat tolerance. Here, we explored the prevalence of this variant in 19 native Chinese cattle (comprised of 404 individuals) to determine its possible association with heat tolerance in Chinese cattle by using PCR and DNA sequencing. The distribution of alleles of NC_037345.1: c.2237A > G: p. His746Arg displays significant geographical differences across native Chinese cattle breeds, consistent with the distribution of indicine and taurine cattle in China. Additionally, the association analysis indicated that the G allele was significantly associated with mean annual temperature (T), relative humidity (RH) and temperature humidity index (THI) (p < .05), suggesting that cattle carrying allele G were distributed in regions with higher T, RH, and THI. In conclusion, our results suggested that the mutation of the TRPM4 gene in Chinese cattle might be a candidate locus associated with heat tolerance.
Collapse
Affiliation(s)
- LuLan Zeng
- College of Life Sciences, Northwest A&F University, Yangling, China
| | - AiXin Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Kaixing Qu
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Jicai Zhang
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Bizhi Huang
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
14
|
Bullerjahn JT, Hanson SM. Extracting thermodynamic properties from van 't Hoff plots with emphasis on temperature-sensing ion channels. Temperature (Austin) 2023; 11:60-71. [PMID: 38577298 PMCID: PMC10989706 DOI: 10.1080/23328940.2023.2265962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/28/2023] [Indexed: 04/06/2024] Open
Abstract
Transient receptor potential (TRP) ion channels are among the most well-studied classes of temperature-sensing molecules. Yet, the molecular mechanism and thermodynamic basis for the temperature sensitivity of TRP channels remains to this day poorly understood. One hypothesis is that the temperature-sensing mechanism can simply be described by a difference in heat capacity between the closed and open channel states. While such a two-state model may be simplistic it nonetheless has descriptive value, in the sense that it can be used to compare overall temperature sensitivity between different channels and mutants. Here, we introduce a mathematical framework based on the two-state model to reliably extract temperature-dependent thermodynamic potentials and heat capacities from measurements of equilibrium constants at different temperatures. Our framework is implemented in an open-source data analysis package that provides a straightforward way to fit both linear and nonlinear van 't Hoff plots, thus avoiding some of the previous, potentially erroneous, assumptions when extracting thermodynamic variables from TRP channel electrophysiology data.
Collapse
Affiliation(s)
- Jakob T. Bullerjahn
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Sonya M. Hanson
- Center for Computational Biology & Center for Computational Mathematics, The Flatiron Institute, New York, NY, USA
| |
Collapse
|
15
|
Jesus RLC, Araujo FA, Alves QL, Dourado KC, Silva DF. Targeting temperature-sensitive transient receptor potential channels in hypertension: far beyond the perception of hot and cold. J Hypertens 2023; 41:1351-1370. [PMID: 37334542 DOI: 10.1097/hjh.0000000000003487] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Transient receptor potential (TRP) channels are nonselective cation channels and participate in various physiological roles. Thus, changes in TRP channel function or expression have been linked to several disorders. Among the many TRP channel subtypes, the TRP ankyrin type 1 (TRPA1), TRP melastatin type 8 (TRPM8), and TRP vanilloid type 1 (TRPV1) channels are temperature-sensitive and recognized as thermo-TRPs, which are expressed in the primary afferent nerve. Thermal stimuli are converted into neuronal activity. Several studies have described the expression of TRPA1, TRPM8, and TRPV1 in the cardiovascular system, where these channels can modulate physiological and pathological conditions, including hypertension. This review provides a complete understanding of the functional role of the opposing thermo-receptors TRPA1/TRPM8/TRPV1 in hypertension and a more comprehensive appreciation of TRPA1/TRPM8/TRPV1-dependent mechanisms involved in hypertension. These channels varied activation and inactivation have revealed a signaling pathway that may lead to innovative future treatment options for hypertension and correlated vascular diseases.
Collapse
Affiliation(s)
- Rafael Leonne C Jesus
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Fênix A Araujo
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Bahia, Brazil
| | - Quiara L Alves
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Keina C Dourado
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Darizy F Silva
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Bahia, Brazil
| |
Collapse
|
16
|
Pertusa M, Solorza J, Madrid R. Molecular determinants of TRPM8 function: key clues for a cool modulation. Front Pharmacol 2023; 14:1213337. [PMID: 37388453 PMCID: PMC10301734 DOI: 10.3389/fphar.2023.1213337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/30/2023] [Indexed: 07/01/2023] Open
Abstract
Cold thermoreceptor neurons detect temperature drops with highly sensitive molecular machinery concentrated in their peripheral free nerve endings. The main molecular entity responsible for cold transduction in these neurons is the thermo-TRP channel TRPM8. Cold, cooling compounds such as menthol, voltage, and osmolality rises activate this polymodal ion channel. Dysregulation of TRPM8 activity underlies several physiopathological conditions, including painful cold hypersensitivity in response to axonal damage, migraine, dry-eye disease, overactive bladder, and several forms of cancer. Although TRPM8 could be an attractive target for treating these highly prevalent diseases, there is still a need for potent and specific modulators potentially suitable for future clinical trials. This goal requires a complete understanding of the molecular determinants underlying TRPM8 activation by chemical and physical agonists, inhibition by antagonists, and the modulatory mechanisms behind its function to guide future and more successful treatment strategies. This review recapitulates information obtained from different mutagenesis approaches that have allowed the identification of specific amino acids in the cavity comprised of the S1-S4 and TRP domains that determine modulation by chemical ligands. In addition, we summarize different studies revealing specific regions within the N- and C-terminus and the transmembrane domain that contribute to cold-dependent TRPM8 gating. We also highlight the latest milestone in the field: cryo-electron microscopy structures of TRPM8, which have provided a better comprehension of the 21 years of extensive research in this ion channel, shedding light on the molecular bases underlying its modulation, and promoting the future rational design of novel drugs to selectively regulate abnormal TRPM8 activity under pathophysiological conditions.
Collapse
Affiliation(s)
- María Pertusa
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Jocelyn Solorza
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, Talca, Chile
| | - Rodolfo Madrid
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| |
Collapse
|
17
|
Yeh F, Jara-Oseguera A, Aldrich RW. Implications of a temperature-dependent heat capacity for temperature-gated ion channels. Proc Natl Acad Sci U S A 2023; 120:e2301528120. [PMID: 37279277 PMCID: PMC10268252 DOI: 10.1073/pnas.2301528120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/26/2023] [Indexed: 06/08/2023] Open
Abstract
Temperature influences dynamics and state-equilibrium distributions in all molecular processes, and only a relatively narrow range of temperatures is compatible with life-organisms must avoid temperature extremes that can cause physical damage or metabolic disruption. Animals evolved a set of sensory ion channels, many of them in the family of transient receptor potential cation channels that detect biologically relevant changes in temperature with remarkable sensitivity. Depending on the specific ion channel, heating or cooling elicits conformational changes in the channel to enable the flow of cations into sensory neurons, giving rise to electrical signaling and sensory perception. The molecular mechanisms responsible for the heightened temperature-sensitivity in these ion channels, as well as the molecular adaptations that make each channel specifically heat- or cold-activated, are largely unknown. It has been hypothesized that a heat capacity difference (ΔCp) between two conformational states of these biological thermosensors can drive their temperature-sensitivity, but no experimental measurements of ΔCp have been achieved for these channel proteins. Contrary to the general assumption that the ΔCp is constant, measurements from soluble proteins indicate that the ΔCp is likely to be a function of temperature. By investigating the theoretical consequences for a linearly temperature-dependent ΔCp on the open-closed equilibrium of an ion channel, we uncover a range of possible channel behaviors that are consistent with experimental measurements of channel activity and that extend beyond what had been generally assumed to be possible for a simple two-state model, challenging long-held assumptions about ion channel gating models at equilibrium.
Collapse
Affiliation(s)
- Frank Yeh
- Institute for Neuroscience, University of Texas at Austin, Austin, TX78712
- Department of Neuroscience, University of Texas at Austin, Austin, TX78712
| | - Andrés Jara-Oseguera
- Institute for Neuroscience, University of Texas at Austin, Austin, TX78712
- Department of Neuroscience, University of Texas at Austin, Austin, TX78712
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX78712
| | - Richard W. Aldrich
- Institute for Neuroscience, University of Texas at Austin, Austin, TX78712
- Department of Neuroscience, University of Texas at Austin, Austin, TX78712
| |
Collapse
|
18
|
Progress in the Structural Basis of thermoTRP Channel Polymodal Gating. Int J Mol Sci 2023; 24:ijms24010743. [PMID: 36614186 PMCID: PMC9821180 DOI: 10.3390/ijms24010743] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
The thermosensory transient receptor potential (thermoTRP) family of ion channels is constituted by several nonselective cation channels that are activated by physical and chemical stimuli functioning as paradigmatic polymodal receptors. Gating of these ion channels is achieved through changes in temperature, osmolarity, voltage, pH, pressure, and by natural or synthetic chemical compounds that directly bind to these proteins to regulate their activity. Given that thermoTRP channels integrate diverse physical and chemical stimuli, a thorough understanding of the molecular mechanisms underlying polymodal gating has been pursued, including the interplay between stimuli and differences between family members. Despite its complexity, recent advances in cryo-electron microscopy techniques are facilitating this endeavor by providing high-resolution structures of these channels in different conformational states induced by ligand binding or temperature that, along with structure-function and molecular dynamics, are starting to shed light on the underlying allosteric gating mechanisms. Because dysfunctional thermoTRP channels play a pivotal role in human diseases such as chronic pain, unveiling the intricacies of allosteric channel gating should facilitate the development of novel drug-based resolving therapies for these disorders.
Collapse
|
19
|
Rosenbaum T, Morales-Lázaro SL. Regulation of ThermoTRP Channels by PIP2 and Cholesterol. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:245-277. [PMID: 36988884 DOI: 10.1007/978-3-031-21547-6_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Transient receptor potential (TRP) ion channels are proteins that are expressed by diverse tissues and that play pivotal functions in physiology. These channels are polymodal and are activated by several stimuli. Among TRPs, some members of this family of channels respond to changes in ambient temperature and are known as thermoTRPs. These proteins respond to heat or cold in the noxious range and some of them to temperatures considered innocuous, as well as to mechanical, osmotic, and/or chemical stimuli. In addition to this already complex ability to respond to different signals, the activity of these ion channels can be fine-tuned by lipids. Two lipids well known to modulate ion channel activity are phosphatidylinositol 4,5-bisphosphate (PIP2) and cholesterol. These lipids can either influence the function of these proteins through direct interaction by binding to a site in the structure of the ion channel or through indirect mechanisms, which can include modifying membrane properties, such as curvature and rigidity, by regulating their expression or by modulating the actions of other molecules or signaling pathways that affect the physiology of ion channels. Here, we summarize the key aspects of the regulation of thermoTRP channels by PIP2 and cholesterol.
Collapse
Affiliation(s)
- Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - Sara L Morales-Lázaro
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
20
|
Zhang L, Li N, Dayananda B, Wang L, Chen H, Cao Y. Genome-Wide Identification and Phylogenetic Analysis of TRP Gene Family Members in Saurian. Animals (Basel) 2022; 12:3593. [PMID: 36552513 PMCID: PMC9774356 DOI: 10.3390/ani12243593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The transient receptor potential plays a critical role in the sensory nervous systems of vertebrates in response to various mechanisms and stimuli, such as environmental temperature. We studied the physiological adaptive evolution of the TRP gene in the saurian family and performed a comprehensive analysis to identify the evolution of the thermo-TRPs channels. All 251 putative TRPs were divided into 6 subfamilies, except TRPN, from the 8 saurian genomes. Multiple characteristics of these genes were analyzed. The results showed that the most conserved proteins of TRP box 1 were located in motif 1, and those of TRP box 2 were located in motif 10. The TRPA and TRPV in saurian tend to be one cluster, as a sister cluster with TRPC, and the TRPM is the root of group I. The TRPM, TRPV, and TRPP were clustered into two clades, and TRPP were organized into TRP PKD1-like and PKD2-like. Segmental duplications mainly occurred in the TRPM subfamily, and tandem duplications only occurred in the TRPV subfamily. There were 15 sites to be under positive selection for TRPA1 and TRPV2 genes. In summary, gene structure, chromosomal location, gene duplication, synteny analysis, and selective pressure at the molecular level provided some new evidence for genetic adaptation to the environment. This result provides a basis for identifying and classifying TRP genes and contributes to further elucidating their potential function in thermal sensors.
Collapse
Affiliation(s)
- Lin Zhang
- School of Health and Nursing, Wuchang University of Technology, Wuhan 430223, China
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Ning Li
- College of Food Science, Nanjing Xiaozhuang University, Nanjing 211171, China
| | - Buddhi Dayananda
- School of Agriculture and Food Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Lihu Wang
- School of Landscape and Ecological Engineering, Hebei University of Engineering, Handan 056038, China
| | - Huimin Chen
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yunpeng Cao
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
| |
Collapse
|
21
|
Dual amplification strategy turns TRPM2 channels into supersensitive central heat detectors. Proc Natl Acad Sci U S A 2022; 119:e2212378119. [PMID: 36409885 PMCID: PMC9881722 DOI: 10.1073/pnas.2212378119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The Ca2+ and ADP ribose (ADPR)-activated cation channel TRPM2 is the closest homolog of the cold sensor TRPM8 but serves as a deep-brain warmth sensor. To unravel the molecular mechanism of heat sensing by the TRPM2 protein, we study here temperature dependence of TRPM2 currents in cell-free membrane patches across ranges of agonist concentrations. We find that channel gating remains strictly agonist-dependent even at 40°C: heating alone or in combination with just Ca2+, just ADPR, Ca2+ + cyclic ADPR, or H2O2 pretreatment only marginally activates TRPM2. For fully liganded TRPM2, pore opening is intrinsically endothermic, due to ~10-fold larger activation enthalpy for opening (~200 kJ/mol) than for closure (~20 kJ/mol). However, the temperature threshold is too high (>40°C) for unliganded but too low (<15°C) for fully liganded channels. Thus, warmth sensitivity around 37°C is restricted to narrow ranges of agonist concentrations. For ADPR, that range matches, but for Ca2+, it exceeds bulk cytosolic values. The supraphysiological [Ca2+] needed for TRPM2 warmth sensitivity is provided by Ca2+ entering through the channel's pore. That positive feedback provides further strong amplification to the TRPM2 temperature response (Q10 ~ 1,000), enabling the TRPM2 protein to autonomously respond to tiny temperature fluctuations around 37°C. These functional data together with published structures suggest a molecular mechanism for opposite temperature dependences of two closely related channel proteins.
Collapse
|
22
|
Plaza‐Cayón A, González‐Muñiz R, Martín‐Martínez M. Mutations of TRPM8 channels: Unraveling the molecular basis of activation by cold and ligands. Med Res Rev 2022; 42:2168-2203. [PMID: 35976012 PMCID: PMC9805079 DOI: 10.1002/med.21920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 01/09/2023]
Abstract
The cation nonselective channel TRPM8 is activated by multiple stimuli, including moderate cold and various chemical compounds (i.e., menthol and icilin [Fig. 1], among others). While research continues growing on the understanding of the physiological involvement of TRPM8 channels and their role in various pathological states, the information available on its activation mechanisms has also increased, supported by mutagenesis and structural studies. This review compiles known information on specific mutations of channel residues and their consequences on channel viability and function. Besides, the comparison of sequence of animals living in different environments, together with chimera and mutagenesis studies are helping to unravel the mechanism of adaptation to different temperatures. The results of mutagenesis studies, grouped by different channel regions, are compared with the current knowledge of TRPM8 structures obtained by cryo-electron microscopy. Trying to make this review self-explicative and highly informative, important residues for TRPM8 function are summarized in a figure, and mutants, deletions and chimeras are compiled in a table, including also the observed effects by different methods of activation and the corresponding references. The information provided by this review may also help in the design of new ligands for TRPM8, an interesting biological target for therapeutic intervention.
Collapse
|
23
|
Sugino S, Inoue K, Kobayashi R, Hirose R, Doi T, Harusato A, Dohi O, Yoshida N, Uchiyama K, Ishikawa T, Takagi T, Yasuda H, Konishi H, Hirai Y, Mizushima K, Naito Y, Tsuji T, Okuda T, Kagawa K, Tominaga M, Itoh Y. Association Between the Cool Temperature-dependent Suppression of Colonic Peristalsis and Transient Receptor Potential Melastatin 8 Activation in Both a Randomized Clinical Trial and an Animal Model. J Neurogastroenterol Motil 2022; 28:693-705. [PMID: 36250375 PMCID: PMC9577569 DOI: 10.5056/jnm21198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/29/2021] [Accepted: 02/20/2022] [Indexed: 12/04/2022] Open
Abstract
Background/Aims Several studies have assessed the effect of cool temperature on colonic peristalsis. Transient receptor potential melastatin 8 (TRPM8) is a temperature-sensitive ion channel activated by mild cooling expressed in the colon. We examined the antispasmodic effect of cool temperature on colonic peristalsis in a prospective, randomized, single-blind trial and based on the video imaging and intraluminal pressure of the proximal colon in rats and TRPM8-deficient mice. Methods In the clinical trial, we randomly assigned a total of 94 patients scheduled to undergo colonoscopy to 2 groups: the mildly cool water (n = 47) and control (n = 47) groups. We used 20 mL of 15°C water for the mildly cool water. The primary outcome was the proportion of subjects with improved peristalsis after treatment. In the rodent proximal colon, we evaluated the intraluminal pressure and performed video imaging of the rodent proximal colon with cool water administration into the colonic lumen. Clinical trial registry website (Trial No. UMIN-CTR; UMIN000030725). Results In the randomized controlled trial, after treatment, the proportion of subjects with no peristalsis with cool water was significantly higher than that in the placebo group (44.7% vs 23.4%; P < 0.05). In the rodent colon model, cool temperature water was associated with a significant decrease in colonic peristalsis through its suppression of the ratio of peak frequency (P < 0.05). Cool temperature-treated TRPM8-deficient mice did not show a reduction in colonic peristalsis compared with wild-type mice. Conclusion For the first time, this study demonstrates that cool temperature-dependent suppression of colonic peristalsis may be associated with TRPM8 activation.
Collapse
Affiliation(s)
- Satoshi Sugino
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Ken Inoue
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Reo Kobayashi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Ryohei Hirose
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Toshifumi Doi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Akihito Harusato
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Osamu Dohi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Naohisa Yoshida
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Kazuhiko Uchiyama
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Takeshi Ishikawa
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Tomohisa Takagi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Hiroaki Yasuda
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Hideyuki Konishi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Yasuko Hirai
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Katsura Mizushima
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshifumi Tsuji
- Department of Gastroenterology and Hepatology, Fukuchiyama City Hospital, Kyoto, Japan
| | - Takashi Okuda
- Department of Gastroenterology and Hepatology, Fukuchiyama City Hospital, Kyoto, Japan
| | - Keizo Kagawa
- Department of Gastroenterology and Hepatology, Fukuchiyama City Hospital, Kyoto, Japan
| | - Makoto Tominaga
- Division of Cell Signaling National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
24
|
Yan C, Wu W, Dong W, Zhu B, Chang J, Lv Y, Yang S, Li JT. Temperature acclimation in hot-spring snakes and the convergence of cold response. Innovation (N Y) 2022; 3:100295. [PMID: 36032194 PMCID: PMC9405097 DOI: 10.1016/j.xinn.2022.100295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/28/2022] [Indexed: 11/05/2022] Open
Abstract
Animals have evolved sophisticated temperature-sensing systems and mechanisms to detect and respond to ambient temperature changes. As a relict species endemic to the Qinghai-Tibet Plateau, hot-spring snake (Thermophis baileyi) survived the dramatic changes in climate that occurred during plateau uplift and ice ages, providing an excellent opportunity to explore the evolution of temperature sensation in ectotherms. Based on distributional information and behavioral experiments, we found that T. baileyi prefer hot-spring habitats and respond more quickly to warmth than other two snakes, suggesting that T. baileyi may evolve an efficient thermal-sensing system. Using high-quality chromosome-level assembly and comparative genomic analysis, we identified cold acclimation genes experiencing convergent acceleration in high-altitude lineages. We also discovered significant evolutionary changes in thermosensation- and thermoregulation-related genes, including the transient receptor potential (TRP) channels. Among these genes, TRPA1 exhibited three species-specific amino acid replacements, which differed from those found in infrared imaging snakes, implying different temperature-sensing molecular strategies. Based on laser-heating experiments, the T. baileyi-specific mutations in TRPA1 resulted in an increase in heat-induced opening probability and thermal sensitivity of the ion channels under the same degree of temperature stimulation, which may help the organism respond to temperature changes more quickly. These results provide insight into the genetic mechanisms underpinning the evolution of temperature-sensing strategies in ectotherms as well as genetic evidence of temperature acclimation in this group. Hot-spring snakes prefer hot-spring habitats on the Qinghai-Tibet Plateau Genetic variation in the snakes contribute to the temperature acclimation Unique mutations in TRPA1 increase thermal sensitivity of the ion channel Different temperature-sensing strategies existed across snakes
Collapse
|
25
|
Cabezas-Bratesco D, Mcgee FA, Colenso CK, Zavala K, Granata D, Carnevale V, Opazo JC, Brauchi SE. Sequence and structural conservation reveal fingerprint residues in TRP channels. eLife 2022; 11:73645. [PMID: 35686986 PMCID: PMC9242649 DOI: 10.7554/elife.73645] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Transient receptor potential (TRP) proteins are a large family of cation-selective channels, surpassed in variety only by voltage-gated potassium channels. Detailed molecular mechanisms governing how membrane voltage, ligand binding, or temperature can induce conformational changes promoting the open state in TRP channels are still a matter of debate. Aiming to unveil distinctive structural features common to the transmembrane domains within the TRP family, we performed phylogenetic reconstruction, sequence statistics, and structural analysis over a large set of TRP channel genes. Here, we report an exceptionally conserved set of residues. This fingerprint is composed of twelve residues localized at equivalent three-dimensional positions in TRP channels from the different subtypes. Moreover, these amino acids are arranged in three groups, connected by a set of aromatics located at the core of the transmembrane structure. We hypothesize that differences in the connectivity between these different groups of residues harbor the apparent differences in coupling strategies used by TRP subgroups.
Collapse
Affiliation(s)
| | - Francisco A Mcgee
- Department of Biology, Temple University, Philadelphia, United States
| | - Charlotte K Colenso
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Kattina Zavala
- Instituto de Ciencias Ambientales y Evolutivas, Universidad Austral de Chile, Valdivia, Chile
| | - Daniele Granata
- Department of Biology, Temple University, Philadelphia, United States
| | | | - Juan C Opazo
- Instituto de Ciencias Ambientales y Evolutivas, Universidad Austral de Chile, Valdivia, Chile
| | | |
Collapse
|
26
|
Zhao C, Xie Y, Xu L, Ye F, Xu X, Yang W, Yang F, Guo J. Structures of a mammalian TRPM8 in closed state. Nat Commun 2022; 13:3113. [PMID: 35662242 PMCID: PMC9166780 DOI: 10.1038/s41467-022-30919-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/25/2022] [Indexed: 11/09/2022] Open
Abstract
Transient receptor potential melastatin 8 (TRPM8) channel is a Ca2+-permeable non-selective cation channel that acts as the primary cold sensor in humans. TRPM8 is also activated by ligands such as menthol, icilin, and phosphatidylinositol 4,5-bisphosphate (PIP2), and desensitized by Ca2+. Here we have determined electron cryo-microscopy structures of mouse TRPM8 in the absence of ligand, and in the presence of Ca2+ and icilin at 2.5–3.2 Å resolution. The ligand-free state TRPM8 structure represents the full-length structure of mammalian TRPM8 channels with a canonical S4-S5 linker and the clearly resolved selectivity filter and outer pore loop. TRPM8 has a short but wide selectivity filter which may account for its permeability to hydrated Ca2+. Ca2+ and icilin bind in the cytosolic-facing cavity of the voltage-sensing-like domain of TRPM8 but induce little conformational change. All the ligand-bound TRPM8 structures adopt the same closed conformation as the ligand-free structure. This study reveals the overall architecture of mouse TRPM8 and the structural basis for its ligand recognition. The mechanism of cold-activated TRPM8 channel activation remains unclear. Here, authors have determined structures of mouse TRPM8 in apo or ligand-bound states, providing insights into the activation of TRPM8 structures in different states.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Yuan Xie
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Lizhen Xu
- Department of Biophysics and Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Fan Ye
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Ximing Xu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, 266003, China
| | - Wei Yang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Fan Yang
- Department of Biophysics and Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China. .,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China. .,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, Zhejiang, 310058, China. .,Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, Zhejiang, 311121, China.
| | - Jiangtao Guo
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China. .,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China. .,Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, Zhejiang, 311121, China. .,State Key Laboratory of Plant Physiology and Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China. .,Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China. .,Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
27
|
Iraci N, Ostacolo C, Medina-Peris A, Ciaglia T, Novoselov AM, Altieri A, Cabañero D, Fernandez-Carvajal A, Campiglia P, Gomez-Monterrey I, Bertamino A, Kurkin AV. In Vitro and In Vivo Pharmacological Characterization of a Novel TRPM8 Inhibitor Chemotype Identified by Small-Scale Preclinical Screening. Int J Mol Sci 2022; 23:2070. [PMID: 35216186 PMCID: PMC8877448 DOI: 10.3390/ijms23042070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/04/2022] [Accepted: 02/11/2022] [Indexed: 11/16/2022] Open
Abstract
Transient receptor potential melastatin type 8 (TRPM8) is a target for the treatment of different physio-pathological processes. While TRPM8 antagonists are reported as potential drugs for pain, cancer, and inflammation, to date only a limited number of chemotypes have been investigated and thus a limited number of compounds have reached clinical trials. Hence there is high value in searching for new TRPM8 antagonistic to broaden clues to structure-activity relationships, improve pharmacological properties and explore underlying molecular mechanisms. To address this, the EDASA Scientific in-house molecular library has been screened in silico, leading to identifying twenty-one potentially antagonist compounds of TRPM8. Calcium fluorometric assays were used to validate the in-silico hypothesis and assess compound selectivity. Four compounds were identified as selective TRPM8 antagonists, of which two were dual-acting TRPM8/TRPV1 modulators. The most potent TRPM8 antagonists (BB 0322703 and BB 0322720) underwent molecular modelling studies to highlight key structural features responsible for drug-protein interaction. The two compounds were also investigated by patch-clamp assays, confirming low micromolar potencies. The most potent compound (BB 0322703, IC50 1.25 ± 0.26 μM) was then profiled in vivo in a cold allodinya model, showing pharmacological efficacy at 30 μM dose. The new chemotypes identified showed remarkable pharmacological properties paving the way to further investigations for drug discovery and pharmacological purposes.
Collapse
Affiliation(s)
- Nunzio Iraci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy;
| | - Carmine Ostacolo
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (C.O.); (I.G.-M.)
| | - Alicia Medina-Peris
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Avenida de la Universidad, 03202 Elche, Spain; (A.M.-P.); (D.C.); (A.F.-C.)
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (P.C.)
| | - Anton M. Novoselov
- Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninsky Gory, 119991 Moscow, Russia; (A.M.N.); (A.A.)
| | - Andrea Altieri
- Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninsky Gory, 119991 Moscow, Russia; (A.M.N.); (A.A.)
- EDASA Scientific srls, Via Stingi 37, 66050 San Salvo, Italy
| | - David Cabañero
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Avenida de la Universidad, 03202 Elche, Spain; (A.M.-P.); (D.C.); (A.F.-C.)
| | - Asia Fernandez-Carvajal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Avenida de la Universidad, 03202 Elche, Spain; (A.M.-P.); (D.C.); (A.F.-C.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (P.C.)
| | - Isabel Gomez-Monterrey
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (C.O.); (I.G.-M.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (P.C.)
| | - Alexander V. Kurkin
- Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninsky Gory, 119991 Moscow, Russia; (A.M.N.); (A.A.)
| |
Collapse
|
28
|
Polymodal Control of TMEM16x Channels and Scramblases. Int J Mol Sci 2022; 23:ijms23031580. [PMID: 35163502 PMCID: PMC8835819 DOI: 10.3390/ijms23031580] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
The TMEM16A/anoctamin-1 calcium-activated chloride channel (CaCC) contributes to a range of vital functions, such as the control of vascular tone and epithelial ion transport. The channel is a founding member of a family of 10 proteins (TMEM16x) with varied functions; some members (i.e., TMEM16A and TMEM16B) serve as CaCCs, while others are lipid scramblases, combine channel and scramblase function, or perform additional cellular roles. TMEM16x proteins are typically activated by agonist-induced Ca2+ release evoked by Gq-protein-coupled receptor (GqPCR) activation; thus, TMEM16x proteins link Ca2+-signalling with cell electrical activity and/or lipid transport. Recent studies demonstrate that a range of other cellular factors—including plasmalemmal lipids, pH, hypoxia, ATP and auxiliary proteins—also control the activity of the TMEM16A channel and its paralogues, suggesting that the TMEM16x proteins are effectively polymodal sensors of cellular homeostasis. Here, we review the molecular pathophysiology, structural biology, and mechanisms of regulation of TMEM16x proteins by multiple cellular factors.
Collapse
|
29
|
Kelava L, Ivić I, Pakai E, Fekete K, Maroti P, Told R, Ujfalusi Z, Garami A. Stereolithography 3D Printing of a Heat Exchanger for Advanced Temperature Control in Wire Myography. Polymers (Basel) 2022; 14:polym14030471. [PMID: 35160461 PMCID: PMC8839612 DOI: 10.3390/polym14030471] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
We report the additive manufacturing of a heat-exchange device that can be used as a cooling accessory in a wire myograph. Wire myography is used for measuring vasomotor responses in small resistance arteries; however, the commercially available devices are not capable of active cooling. Here, we critically evaluated a transparent resin material, in terms of mechanical, structural, and thermal behavior. Tensile strength tests (67.66 ± 1.31 MPa), Charpy impact strength test (20.70 ± 2.30 kJ/m2), and Shore D hardness measurements (83.0 ± 0.47) underlined the mechanical stability of the material, supported by digital microscopy, which revealed a glass-like structure. Differential scanning calorimetry with thermogravimetry analysis and thermal conductivity measurements showed heat stability until ~250 °C and effective heat insulation. The 3D-printed heat exchanger was tested in thermophysiology experiments measuring the vasomotor responses of rat tail arteries at different temperatures (13, 16, and 36 °C). The heat-exchange device was successfully used as an accessory of the wire myograph system to cool down the experimental chambers and steadily maintain the targeted temperatures. We observed temperature-dependent differences in the vasoconstriction induced by phenylephrine and KCl. In conclusion, the transparent resin material can be used in additive manufacturing of heat-exchange devices for biomedical research, such as wire myography. Our animal experiments underline the importance of temperature-dependent physiological mechanisms, which should be further studied to understand the background of the thermal changes and their consequences.
Collapse
Affiliation(s)
- Leonardo Kelava
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, H-7624 Pecs, Hungary
| | - Ivan Ivić
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, H-7624 Pecs, Hungary
| | - Eszter Pakai
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, H-7624 Pecs, Hungary
| | - Kata Fekete
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, H-7624 Pecs, Hungary
| | - Peter Maroti
- Medical Simulation Education Center, Medical School, University of Pecs, H-7624 Pecs, Hungary
- 3D Printing and Visualization Center, University of Pecs, H-7624 Pecs, Hungary
| | - Roland Told
- Medical Simulation Education Center, Medical School, University of Pecs, H-7624 Pecs, Hungary
- 3D Printing and Visualization Center, University of Pecs, H-7624 Pecs, Hungary
| | - Zoltan Ujfalusi
- Department of Biophysics, Medical School, University of Pecs, H-7624 Pecs, Hungary
| | - Andras Garami
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, H-7624 Pecs, Hungary
| |
Collapse
|
30
|
Phan TX, Ton HT, Gulyás H, Pórszász R, Tóth A, Russo R, Kay MW, Sahibzada N, Ahern GP. TRPV1 in arteries enables a rapid myogenic tone. J Physiol 2022; 600:1651-1666. [PMID: 35020949 PMCID: PMC8976781 DOI: 10.1113/jp281873] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/11/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS We explored the physiological role of TRPV1 in vascular smooth muscle. TRPV1 antagonists dilated skeletal muscle arterioles both ex vivo and in vivo, increased coronary perfusion and decreased systemic blood pressure. Stretch of arteriolar myocytes and increases in intraluminal pressure in arteries triggered rapid Ca2+ signaling and vasoconstriction respectively. Pharmacologic and/or genetic disruption of TRPV1 significantly inhibited the magnitude and rate of these responses. Furthermore, disrupting TRPV1 blunted the rapid vasodilation evoked by arterial constriction. Pharmacological experiments identified key roles for phospholipase C and protein kinase C, combined with temperature, in TRPV1-dependent arterial tone. These results show that TRPV1 in arteriolar myocytes dynamically regulates myogenic tone and blood flow in the heart and skeletal muscle. ABSTRACT Arterioles maintain blow flow by adjusting their diameter in response to changes in local blood pressure. In this process called the myogenic response, a vascular smooth muscle mechanosensor controls tone predominantly through altering the membrane potential. In general, myogenic responses occur slowly (minutes). In the heart and skeletal muscle, however, tone is activated rapidly (tens of seconds) and terminated by brief (100 ms) arterial constrictions. Previously, we identified extensive expression of TRPV1 in the smooth muscle of arterioles supplying skeletal muscle, heart and fat. Here we reveal a critical role for TRPV1 in the rapid myogenic tone of these tissues. TRPV1 antagonists dilated skeletal muscle arterioles in vitro and in vivo, increased coronary flow in isolated hearts, and transiently decreased blood pressure. All of these pharmacologic effects were abolished by genetic disruption of TRPV1. Stretch of isolated vascular smooth muscle cells or raised intravascular pressure in arteries triggered Ca2+ signaling and vasoconstriction. The majority of these stretch-responses were TRPV1-mediated, with the remaining tone being inhibited by the TRPM4 antagonist, 9-phenantrol. Notably, tone developed more quickly in arteries from wild-type compared with TRPV1-null mice. Furthermore, the immediate vasodilation following brief constriction of arterioles depended on TRPV1, consistent with a rapid deactivation of TRPV1. Pharmacologic experiments revealed that membrane stretch activates phospholipase C/protein kinase C signaling combined with heat to activate TRPV1, and in turn, L-type Ca2+ channels. These results suggest a critical role, for TRPV1 in the dynamic regulation of myogenic tone and blood flow in the heart and skeletal muscle. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Thieu X Phan
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, USA.,Department of Biology, Vinh University, Vinh City, Vietnam
| | - Hoai T Ton
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, USA.,Department of Biology, Vinh University, Vinh City, Vietnam
| | - Hajnalka Gulyás
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Doctoral School of Pharmaceutical Sciences, Debrecen, Hungary
| | - Róbert Pórszász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Doctoral School of Pharmaceutical Sciences, Debrecen, Hungary
| | - Attila Tóth
- Division of Clinical Physiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Rebekah Russo
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Matthew W Kay
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Niaz Sahibzada
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, USA
| | - Gerard P Ahern
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, USA
| |
Collapse
|
31
|
Thermodynamic and structural basis of temperature-dependent gating in TRP channels. Biochem Soc Trans 2021; 49:2211-2219. [PMID: 34623379 DOI: 10.1042/bst20210301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 11/17/2022]
Abstract
Living organisms require detecting the environmental thermal clues for survival, allowing them to avoid noxious stimuli or find prey moving in the dark. In mammals, the Transient Receptor Potential ion channels superfamily is constituted by 27 polymodal receptors whose activity is controlled by small ligands, peptide toxins, protons and voltage. The thermoTRP channels subgroup exhibits unparalleled temperature dependence -behaving as heat and cold sensors. Functional studies have dissected their biophysical features in detail, and the advances of single-particle Cryogenic Electron microscopy provided the structural framework required to propose detailed channel gating mechanisms. However, merging structural and functional evidence for temperature-driven gating of thermoTRP channels has been a hard nut to crack, remaining an open question nowadays. Here we revisit the highlights on the study of heat and cold sensing in thermoTRP channels in the light of the structural data that has emerged during recent years.
Collapse
|
32
|
Rueda-Ruzafa L, Herrera-Pérez S, Campos-Ríos A, Lamas JA. Are TREK Channels Temperature Sensors? Front Cell Neurosci 2021; 15:744702. [PMID: 34690704 PMCID: PMC8526543 DOI: 10.3389/fncel.2021.744702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022] Open
Abstract
Internal human body normal temperature fluctuates between 36.5 and 37.5°C and it is generally measured in the oral cavity. Interestingly, most electrophysiological studies on the functioning of ion channels and their role in neuronal behavior are carried out at room temperature, which usually oscillates between 22 and 24°C, even when thermosensitive channels are studied. We very often forget that if the core of the body reached that temperature, the probability of death from cardiorespiratory arrest would be extremely high. Does this mean that we are studying ion channels in dying neurons? Thousands of electrophysiological experiments carried out at these low temperatures suggest that most neurons tolerate this aggression quite well, at least for the duration of the experiments. This also seems to happen with ion channels, although studies at different temperatures indicate large changes in both, neuron and channel behavior. It is known that many chemical, physical and therefore physiological processes, depend to a great extent on body temperature. Temperature clearly affects the kinetics of numerous events such as chemical reactions or conformational changes in proteins but, what if these proteins constitute ion channels and these channels are specifically designed to detect changes in temperature? In this review, we discuss the importance of the potassium channels of the TREK subfamily, belonging to the recently discovered family of two-pore domain channels, in the transduction of thermal sensitivity in different cell types.
Collapse
Affiliation(s)
- Lola Rueda-Ruzafa
- CINBIO, Laboratory of Neuroscience, University of Vigo, Vigo, Spain.,Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), Vigo, Spain
| | - Salvador Herrera-Pérez
- CINBIO, Laboratory of Neuroscience, University of Vigo, Vigo, Spain.,Grupo de Neurofisiología Experimental y Circuitos Neuronales, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Ana Campos-Ríos
- CINBIO, Laboratory of Neuroscience, University of Vigo, Vigo, Spain.,Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), Vigo, Spain
| | - J A Lamas
- CINBIO, Laboratory of Neuroscience, University of Vigo, Vigo, Spain.,Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), Vigo, Spain
| |
Collapse
|
33
|
Constitutive Phosphorylation as a Key Regulator of TRPM8 Channel Function. J Neurosci 2021; 41:8475-8493. [PMID: 34446569 DOI: 10.1523/jneurosci.0345-21.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/04/2021] [Accepted: 08/13/2021] [Indexed: 11/21/2022] Open
Abstract
In mammals, environmental cold sensing conducted by peripheral cold thermoreceptor neurons mostly depends on TRPM8, an ion channel that has evolved to become the main molecular cold transducer. This TRP channel is activated by cold, cooling compounds, such as menthol, voltage, and rises in osmolality. TRPM8 function is regulated by kinase activity that phosphorylates the channel under resting conditions. However, which specific residues, how this post-translational modification modulates TRPM8 activity, and its influence on cold sensing are still poorly understood. By mass spectrometry, we identified four serine residues within the N-terminus (S26, S29, S541, and S542) constitutively phosphorylated in the mouse ortholog. TRPM8 function was examined by Ca2+ imaging and patch-clamp recordings, revealing that treatment with staurosporine, a kinase inhibitor, augmented its cold- and menthol-evoked responses. S29A mutation is sufficient to increase TRPM8 activity, suggesting that phosphorylation of this residue is a central molecular determinant of this negative regulation. Biophysical and total internal reflection fluorescence-based analysis revealed a dual mechanism in the potentiated responses of unphosphorylated TRPM8: a shift in the voltage activation curve toward more negative potentials and an increase in the number of active channels at the plasma membrane. Importantly, basal kinase activity negatively modulates TRPM8 function at cold thermoreceptors from male and female mice, an observation accounted for by mathematical modeling. Overall, our findings suggest that cold temperature detection could be rapidly and reversibly fine-tuned by controlling the TRPM8 basal phosphorylation state, a mechanism that acts as a dynamic molecular brake of this thermo-TRP channel function in primary sensory neurons.SIGNIFICANCE STATEMENT Post-translational modifications are one of the main molecular mechanisms involved in adjusting the sensitivity of sensory ion channels to changing environmental conditions. Here we show, for the first time, that constitutive phosphorylation of the well-conserved serine 29 within the N-terminal domain negatively modulates TRPM8 channel activity, reducing its activation by agonists and decreasing the number of active channels at the plasma membrane. Basal phosphorylation of TRPM8 acts as a key regulator of its function as the main cold-transduction channel, significantly contributing to the net response of primary sensory neurons to temperature reductions. This reversible and dynamic modulatory mechanism opens new opportunities to regulate TRPM8 function in pathologic conditions where this thermo-TRP channel plays a critical role.
Collapse
|
34
|
Csernyus B, Szabó Á, Fiáth R, Zátonyi A, Lázár C, Pongrácz A, Fekete Z. A multimodal, implantable sensor array and measurement system to investigate the suppression of focal epileptic seizure using hypothermia. J Neural Eng 2021; 18. [PMID: 34280911 DOI: 10.1088/1741-2552/ac15e6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/19/2021] [Indexed: 11/12/2022]
Abstract
Objective.Local cooling of the brain as a therapeutic intervention is a promising alternative for patients with epilepsy who do not respond to medication.In vitroandin vivostudies have demonstrated the seizure-suppressing effect of local cooling in various animal models. In our work, focal brain cooling in a bicuculline induced epilepsy model in rats is demonstrated and evaluated using a multimodal micro-electrocorticography (microECoG) device.Approach.We designed and experimentally tested a novel polyimide-based sensor array capable of recording microECoG and temperature signals concurrently from the cortical surface of rats. The effect of cortical cooling after seizure onset was evaluated using 32 electrophysiological sites and eight temperature sensing elements covering the brain hemisphere, where injection of the epileptic drug was performed. The focal cooling of the cortex right above the injection site was accomplished using a miniaturized Peltier chip combined with a heat pipe to transfer heat. Control of cooling and collection of sensor data was provided by a custom designed Arduino based electronic board. We tested the experimental setup using an agar gel modelin vitro, and thenin vivoin Wistar rats.Main results.Spatial variation of temperature during the Peltier controlled cooling was evaluated through calibrated, on-chip platinum temperature sensors. We found that frequency of epileptic discharges was not substantially reduced by cooling the cortical surface to 30 °C, but was suppressed efficiently at temperature values around 20 °C. The multimodal array revealed that seizure-like ictal events far from the focus and not exposed to high drop in temperature can be also inhibited at an extent like the directly cooled area.Significance.Our results imply that not only the absolute drop in temperature determines the efficacy of seizure suppression, and distant cortical areas not directly cooled can be influenced.
Collapse
Affiliation(s)
- B Csernyus
- Research Group for Implantable Microsystems, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Á Szabó
- Research Group for Implantable Microsystems, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.,Roska Tamás Interdisciplinary Doctoral School, Pázmány Péter Catholic University, Budapest, Hungary
| | - R Fiáth
- Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Budapest, Hungary
| | - A Zátonyi
- Research Group for Implantable Microsystems, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - C Lázár
- Microsystems Laboratory, Institute of Technical Physics and Material Sciences, Center for Energy Research, Budapest, Hungary
| | - A Pongrácz
- Research Group for Implantable Microsystems, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Z Fekete
- Research Group for Implantable Microsystems, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| |
Collapse
|
35
|
Becker V, Hui X, Nalbach L, Ampofo E, Lipp P, Menger MD, Laschke MW, Gu Y. Linalool inhibits the angiogenic activity of endothelial cells by downregulating intracellular ATP levels and activating TRPM8. Angiogenesis 2021; 24:613-630. [PMID: 33655414 PMCID: PMC8292279 DOI: 10.1007/s10456-021-09772-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 02/10/2021] [Indexed: 12/29/2022]
Abstract
Angiogenesis crucially contributes to various diseases, such as cancer and diabetic retinopathy. Hence, anti-angiogenic therapy is considered as a powerful strategy against these diseases. Previous studies reported that the acyclic monoterpene linalool exhibits anticancer, anti-inflammatory and anti-oxidative activity. However, the effects of linalool on angiogenesis still remain elusive. Therefore, we investigated the action of (3R)-(-)-linalool, a main enantiomer of linalool, on the angiogenic activity of human dermal microvascular endothelial cells (HDMECs) by a panel of angiogenesis assays. Non-cytotoxic doses of linalool significantly inhibited HDMEC proliferation, migration, tube formation and spheroid sprouting. Linalool also suppressed the vascular sprouting from rat aortic rings. In addition, Matrigel plugs containing linalool exhibited a significantly reduced microvessel density 7 days after implantation into BALB/c mice. Mechanistic analyses revealed that linalool promotes the phosphorylation of extracellular signal-regulated kinase (ERK), downregulates the intracellular level of adenosine triphosphate (ATP) and activates the transient receptor potential cation channel subfamily M (melastatin) member (TRPM)8 in HDMECs. Inhibition of ERK signaling, supplementation of ATP and blockade of TRPM8 significantly counteracted linalool-suppressed HDMEC spheroid sprouting. Moreover, ATP supplementation completely reversed linalool-induced ERK phosphorylation. In addition, linalool-induced ERK phosphorylation inhibited the expression of bone morphogenetic protein (BMP)-2 and linalool-induced TRPM8 activation caused the inhibition of β1 integrin/focal adhesion kinase (FAK) signaling. These findings indicate an anti-angiogenic effect of linalool, which is mediated by downregulating intracellular ATP levels and activating TRPM8.
Collapse
Affiliation(s)
- Vivien Becker
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Saarland, Germany
| | - Xin Hui
- Molecular Cell Biology, Research Center for Molecular Imaging and Screening, Medical Faculty, Saarland University, 66421, Homburg, Saarland, Germany
| | - Lisa Nalbach
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Saarland, Germany
| | - Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Saarland, Germany
| | - Peter Lipp
- Molecular Cell Biology, Research Center for Molecular Imaging and Screening, Medical Faculty, Saarland University, 66421, Homburg, Saarland, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Saarland, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Saarland, Germany
| | - Yuan Gu
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Saarland, Germany.
| |
Collapse
|
36
|
Gavel EH, Hawke KV, Bentley DJ, Logan-Sprenger HM. Menthol Mouth Rinsing Is More Than Just a Mouth Wash-Swilling of Menthol to Improve Physiological Performance. Front Nutr 2021; 8:691695. [PMID: 34307438 PMCID: PMC8292615 DOI: 10.3389/fnut.2021.691695] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/10/2021] [Indexed: 01/13/2023] Open
Abstract
Interventions that solely act on the central nervous system (CNS) are gaining considerable interest, particularly products consumed through the oral cavity. The oropharyngeal cavity contains a wide array of receptors that respond to sweet, bitter, and cold tastants, all of which have been shown to improve physiological performance. Of late, the ergogenic benefits of carbohydrate (CHO) and caffeine (CAF) mouth rinsings (MRs) have been widely studied; however, less is known about menthol (MEN). That the physiological state and environmental conditions impact the response each product has is increasingly recognized. While the effects of CHO and CAF MRs have been thoroughly studied in both hot and thermoneutral conditions, less is known about MEN as it has only been studied in hot environments. As such, this review summarizes the current knowledge regarding the MEN MR and exercise modality, frequency of the mouth rinse, and mouth rinse duration and compares two different types of study designs: time trials vs. time to exhaustion (TTE).
Collapse
Affiliation(s)
- Erica H Gavel
- Faculty of Science, Ontario Tech University, Oshawa, ON, Canada
| | - Kierstyn V Hawke
- Faculty of Health Science, Ontario Tech University, Oshawa, ON, Canada
| | - David J Bentley
- Faculty of Health Science, Ontario Tech University, Oshawa, ON, Canada
| | - Heather M Logan-Sprenger
- Faculty of Science, Ontario Tech University, Oshawa, ON, Canada.,Faculty of Health Science, Ontario Tech University, Oshawa, ON, Canada.,Canadian Sport Institute Ontario, Toronto, ON, Canada
| |
Collapse
|
37
|
Nadezhdin KD, Neuberger A, Trofimov YA, Krylov NA, Sinica V, Kupko N, Vlachova V, Zakharian E, Efremov RG, Sobolevsky AI. Structural mechanism of heat-induced opening of a temperature-sensitive TRP channel. Nat Struct Mol Biol 2021; 28:564-572. [PMID: 34239124 PMCID: PMC8283911 DOI: 10.1038/s41594-021-00615-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Numerous physiological functions rely on distinguishing temperature through temperature-sensitive transient receptor potential channels (thermo-TRPs). Although the function of thermo-TRPs has been studied extensively, structural determination of their heat- and cold-activated states has remained a challenge. Here, we present cryo-EM structures of the nanodisc-reconstituted wild-type mouse TRPV3 in three distinct conformations: closed, heat-activated sensitized and open states. The heat-induced transformations of TRPV3 are accompanied by changes in the secondary structure of the S2-S3 linker and the N and C termini and represent a conformational wave that links these parts of the protein to a lipid occupying the vanilloid binding site. State-dependent differences in the behavior of bound lipids suggest their active role in thermo-TRP temperature-dependent gating. Our structural data, supported by physiological recordings and molecular dynamics simulations, provide an insight for understanding the molecular mechanism of temperature sensing.
Collapse
Affiliation(s)
- Kirill D. Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA
| | - Arthur Neuberger
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA
| | - Yuri A. Trofimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia,National Research Nuclear University Moscow Engineering Physics Institute, Moscow, Russia,Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| | - Nikolay A. Krylov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia,National Research University Higher School of Economics, Moscow, Russia
| | - Viktor Sinica
- Department of Cellular Neurophysiology, Institute of Physiology Czech Academy of Sciences, Prague, Czech Republic
| | - Nikita Kupko
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA
| | - Viktorie Vlachova
- Department of Cellular Neurophysiology, Institute of Physiology Czech Academy of Sciences, Prague, Czech Republic
| | - Eleonora Zakharian
- Department of Cancer Biology & Pharmacology, University of Illinois College of Medicine, Peoria, Illinois, USA
| | - Roman G. Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia,Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia,National Research University Higher School of Economics, Moscow, Russia
| | - Alexander I. Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA,Correspondence and requests for materials should be addressed to A.I.S. (; Tel: 212-305-4249)
| |
Collapse
|
38
|
Heat-dependent opening of TRPV1 in the presence of capsaicin. Nat Struct Mol Biol 2021; 28:554-563. [PMID: 34239123 PMCID: PMC8335751 DOI: 10.1038/s41594-021-00616-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023]
Abstract
Transient receptor potential vanilloid member 1 (TRPV1) is a Ca2+-permeable cation channel that serves as the primary heat and capsaicin sensor in humans. Using cryo-EM, we have determined the structures of apo and capsaicin-bound full-length rat TRPV1 reconstituted into lipid nanodiscs over a range of temperatures. This has allowed us to visualize the noxious heat-induced opening of TRPV1 in the presence of capsaicin. Notably, noxious heat-dependent TRPV1 opening comprises stepwise conformational transitions. Global conformational changes across multiple subdomains of TRPV1 are followed by the rearrangement of the outer pore, leading to gate opening. Solvent-accessible surface area analyses and functional studies suggest that a subset of residues form an interaction network that is directly involved in heat sensing. Our study provides a glimpse of the molecular principles underlying noxious physical and chemical stimuli sensing by TRPV1, which can be extended to other thermal sensing ion channels.
Collapse
|
39
|
Cao E. Structural mechanisms of transient receptor potential ion channels. J Gen Physiol 2021; 152:133640. [PMID: 31972006 PMCID: PMC7054860 DOI: 10.1085/jgp.201811998] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 01/03/2020] [Indexed: 12/26/2022] Open
Abstract
Transient receptor potential (TRP) ion channels are evolutionarily ancient sensory proteins that detect and integrate a wide range of physical and chemical stimuli. TRP channels are fundamental for numerous biological processes and are therefore associated with a multitude of inherited and acquired human disorders. In contrast to many other major ion channel families, high-resolution structures of TRP channels were not available before 2013. Remarkably, however, the subsequent “resolution revolution” in cryo-EM has led to an explosion of TRP structures in the last few years. These structures have confirmed that TRP channels assemble as tetramers and resemble voltage-gated ion channels in their overall architecture. But beyond the relatively conserved transmembrane core embedded within the lipid bilayer, each TRP subtype appears to be endowed with a unique set of soluble domains that may confer diverse regulatory mechanisms. Importantly, TRP channel structures have revealed sites and mechanisms of action of numerous synthetic and natural compounds, as well as those for endogenous ligands such as lipids, Ca2+, and calmodulin. Here, I discuss these recent findings with a particular focus on the conserved transmembrane region and how these structures may help to rationally target this important class of ion channels for the treatment of numerous human conditions.
Collapse
Affiliation(s)
- Erhu Cao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
| |
Collapse
|
40
|
Thermoregulatory Response to Cold at Various Levels of Activation of Peripheral TRPA1 Ion Channel. Bull Exp Biol Med 2021; 170:420-424. [PMID: 33713225 DOI: 10.1007/s10517-021-05079-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Indexed: 10/21/2022]
Abstract
The effect of TRPA1-ion channel on thermoregulatory responses depending on the level of its activity was studied in Wistar rats. To activate the TRPA1 ion channel localized in the skin, its agonist allyl isothiocyanate (AITC) was used in different concentrations (0.04, 0.4, 1, and 2.5%). Low concentration of AITC (0.04%) enhanced and high concentrations (1 and 2.5%), on the contrary, inhibited cold-defense responses (decreased their magnitude and led to their later initiation due to an increase in temperature thresholds). With an increase in TRPA1 activation, the increase in temperature thresholds (afferent link) was ahead of the decrease in the magnitude of responses (efferent link), which can attest to different sensitivity of these processes to TRPA1 activation.
Collapse
|
41
|
Structural basis for promiscuous action of monoterpenes on TRP channels. Commun Biol 2021; 4:293. [PMID: 33674682 PMCID: PMC7935860 DOI: 10.1038/s42003-021-01776-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 01/22/2021] [Indexed: 01/31/2023] Open
Abstract
Monoterpenes are major constituents of plant-derived essential oils and have long been widely used for therapeutic and cosmetic applications. The monoterpenes menthol and camphor are agonists or antagonists for several TRP channels such as TRPM8, TRPV1, TRPV3 and TRPA1. However, which regions within TRPV1 and TRPV3 confer sensitivity to monoterpenes or other synthesized chemicals such as 2-APB are unclear. In this study we identified conserved arginine and glycine residues in the linker between S4 and S5 that are related to the action of these chemicals and validated these findings in molecular dynamics simulations. The involvement of these amino acids differed between TRPV3 and TRPV1 for chemical-induced and heat-evoked activation. These findings provide the basis for characterization of physiological function and biophysical properties of ion channels.
Collapse
|
42
|
Henao JC, Grismaldo A, Barreto A, Rodríguez-Pardo VM, Mejía-Cruz CC, Leal-Garcia E, Pérez-Núñez R, Rojas P, Latorre R, Carvacho I, Torres YP. TRPM8 Channel Promotes the Osteogenic Differentiation in Human Bone Marrow Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:592946. [PMID: 33614639 PMCID: PMC7890257 DOI: 10.3389/fcell.2021.592946] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/05/2021] [Indexed: 11/29/2022] Open
Abstract
Various families of ion channels have been characterized in mesenchymal stem cells (MSCs), including some members of transient receptor potential (TRP) channels family. TRP channels are involved in critical cellular processes as differentiation and cell proliferation. Here, we analyzed the expression of TRPM8 channel in human bone marrow MSCs (hBM-MSCs), and its relation with osteogenic differentiation. Patch-clamp recordings showed that hBM-MSCs expressed outwardly rectifying currents which were increased by exposure to 500 μM menthol and were partially inhibited by 10 μM of BCTC, a TRPM8 channels antagonist. Additionally, we have found the expression of TRPM8 by RT-PCR and western blot. We also explored the TRPM8 localization in hBM-MSCs by immunofluorescence using confocal microscopy. Remarkably, hBM-MSCs treatment with 100 μM of menthol or 10 μM of icilin, TRPM8 agonists, increases osteogenic differentiation. Conversely, 20 μM of BCTC, induced a decrease of osteogenic differentiation. These results suggest that TRPM8 channels are functionally active in hBM-MSCs and have a role in cell differentiation.
Collapse
Affiliation(s)
- Juan C Henao
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Adriana Grismaldo
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alfonso Barreto
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Viviana M Rodríguez-Pardo
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Claudia Camila Mejía-Cruz
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Efrain Leal-Garcia
- Departamento de Ortopedia y Traumatología, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Patricio Rojas
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ingrid Carvacho
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, Talca, Chile
| | - Yolima P Torres
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
43
|
A Tangled Threesome: Circadian Rhythm, Body Temperature Variations, and the Immune System. BIOLOGY 2021; 10:biology10010065. [PMID: 33477463 PMCID: PMC7829919 DOI: 10.3390/biology10010065] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 12/14/2022]
Abstract
Simple Summary In mammals, including humans, the body temperature displays a circadian rhythm and is maintained within a narrow range to facilitate the optimal functioning of physiological processes. Body temperature increases during the daytime and decreases during the nighttime thus influencing the expression of the molecular clock and the clock-control genes such as immune genes. An increase in body temperature (daytime, or fever) also prepares the organism to fight aggression by promoting the activation, function, and delivery of immune cells. Many factors may affect body temperature level and rhythm, including environment, age, hormones, or treatment. The disruption of the body temperature is associated with many kinds of diseases and their severity, thus supporting the assumed association between body temperature rhythm and immune functions. Recent studies using complex analysis suggest that circadian rhythm may change in all aspects (level, period, amplitude) and may be predictive of good or poor outcomes. The monitoring of body temperature is an easy tool to predict outcomes and maybe guide future studies in chronotherapy. Abstract The circadian rhythm of the body temperature (CRBT) is a marker of the central biological clock that results from multiple complex biological processes. In mammals, including humans, the body temperature displays a strict circadian rhythm and has to be maintained within a narrow range to allow optimal physiological functions. There is nowadays growing evidence on the role of the temperature circadian rhythm on the expression of the molecular clock. The CRBT likely participates in the phase coordination of circadian timekeepers in peripheral tissues, thus guaranteeing the proper functioning of the immune system. The disruption of the CRBT, such as fever, has been repeatedly described in diseases and likely reflects a physiological process to activate the molecular clock and trigger the immune response. On the other hand, temperature circadian disruption has also been described as associated with disease severity and thus may mirror or contribute to immune dysfunction. The present review aims to characterize the potential implication of the temperature circadian rhythm on the immune response, from molecular pathways to diseases. The origin of CRBT and physiological changes in body temperature will be mentioned. We further review the immune biological effects of temperature rhythmicity in hosts, vectors, and pathogens. Finally, we discuss the relationship between circadian disruption of the body temperature and diseases and highlight the emerging evidence that CRBT monitoring would be an easy tool to predict outcomes and guide future studies in chronotherapy.
Collapse
|
44
|
Gualdani R, Yuan JH, Effraim PR, Di Stefano G, Truini A, Cruccu G, Dib-Hajj SD, Gailly P, Waxman SG. Trigeminal Neuralgia TRPM8 Mutation: Enhanced Activation, Basal [Ca 2+] i and Menthol Response. NEUROLOGY-GENETICS 2021; 7:e550. [PMID: 33977138 PMCID: PMC8105906 DOI: 10.1212/nxg.0000000000000550] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/09/2020] [Indexed: 01/20/2023]
Abstract
Objective To assess the functional effects of a variant, c.89 G > A (p.Arg30Gln), in the transient receptor potential melastatin 8 (TRPM8) cold-sensing, nonselective cation channel, which we have previously identified in a patient with familial trigeminal neuralgia. Methods We carried out Ca2+ imaging and whole-cell patch-clamp recording. Results The TRPM8 mutation enhances channel activation, increases basal current amplitude and intracellular [Ca2+] in cells carrying the mutant channel, and enhances the response to menthol. Conclusions We propose that Arg30Gln confers gain-of-function attributes on TRPM8, which contribute to pathogenesis of trigeminal neuralgia in patients carrying this mutation.
Collapse
Affiliation(s)
- Roberta Gualdani
- Institute of Neuroscience (R.G., P.G.), Université catholique de Louvain, B-1200 Brussels, Belgium; Department of Neurology (J.-H.Y., S.D.D.-H., S.G.W.), Yale School of Medicine, New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; Department of Anaesthesiology (P.R.E.), Yale School of Medicine New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; and Department of Human Neuroscience (G.D.S., A.T., G.C.), Sapienza University, Rome, Italy
| | - Jun-Hui Yuan
- Institute of Neuroscience (R.G., P.G.), Université catholique de Louvain, B-1200 Brussels, Belgium; Department of Neurology (J.-H.Y., S.D.D.-H., S.G.W.), Yale School of Medicine, New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; Department of Anaesthesiology (P.R.E.), Yale School of Medicine New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; and Department of Human Neuroscience (G.D.S., A.T., G.C.), Sapienza University, Rome, Italy
| | - Philip R Effraim
- Institute of Neuroscience (R.G., P.G.), Université catholique de Louvain, B-1200 Brussels, Belgium; Department of Neurology (J.-H.Y., S.D.D.-H., S.G.W.), Yale School of Medicine, New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; Department of Anaesthesiology (P.R.E.), Yale School of Medicine New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; and Department of Human Neuroscience (G.D.S., A.T., G.C.), Sapienza University, Rome, Italy
| | - Giulia Di Stefano
- Institute of Neuroscience (R.G., P.G.), Université catholique de Louvain, B-1200 Brussels, Belgium; Department of Neurology (J.-H.Y., S.D.D.-H., S.G.W.), Yale School of Medicine, New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; Department of Anaesthesiology (P.R.E.), Yale School of Medicine New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; and Department of Human Neuroscience (G.D.S., A.T., G.C.), Sapienza University, Rome, Italy
| | - Andrea Truini
- Institute of Neuroscience (R.G., P.G.), Université catholique de Louvain, B-1200 Brussels, Belgium; Department of Neurology (J.-H.Y., S.D.D.-H., S.G.W.), Yale School of Medicine, New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; Department of Anaesthesiology (P.R.E.), Yale School of Medicine New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; and Department of Human Neuroscience (G.D.S., A.T., G.C.), Sapienza University, Rome, Italy
| | - Giorgio Cruccu
- Institute of Neuroscience (R.G., P.G.), Université catholique de Louvain, B-1200 Brussels, Belgium; Department of Neurology (J.-H.Y., S.D.D.-H., S.G.W.), Yale School of Medicine, New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; Department of Anaesthesiology (P.R.E.), Yale School of Medicine New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; and Department of Human Neuroscience (G.D.S., A.T., G.C.), Sapienza University, Rome, Italy
| | - Sulayman D Dib-Hajj
- Institute of Neuroscience (R.G., P.G.), Université catholique de Louvain, B-1200 Brussels, Belgium; Department of Neurology (J.-H.Y., S.D.D.-H., S.G.W.), Yale School of Medicine, New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; Department of Anaesthesiology (P.R.E.), Yale School of Medicine New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; and Department of Human Neuroscience (G.D.S., A.T., G.C.), Sapienza University, Rome, Italy
| | - Philippe Gailly
- Institute of Neuroscience (R.G., P.G.), Université catholique de Louvain, B-1200 Brussels, Belgium; Department of Neurology (J.-H.Y., S.D.D.-H., S.G.W.), Yale School of Medicine, New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; Department of Anaesthesiology (P.R.E.), Yale School of Medicine New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; and Department of Human Neuroscience (G.D.S., A.T., G.C.), Sapienza University, Rome, Italy
| | - Stephen G Waxman
- Institute of Neuroscience (R.G., P.G.), Université catholique de Louvain, B-1200 Brussels, Belgium; Department of Neurology (J.-H.Y., S.D.D.-H., S.G.W.), Yale School of Medicine, New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; Department of Anaesthesiology (P.R.E.), Yale School of Medicine New Haven, CT and Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT; and Department of Human Neuroscience (G.D.S., A.T., G.C.), Sapienza University, Rome, Italy
| |
Collapse
|
45
|
The Role of Thermosensitive Ion Channels in Mammalian Thermoregulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:355-370. [DOI: 10.1007/978-981-16-4254-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
46
|
VDAC Gating Thermodynamics, but Not Gating Kinetics, Are Virtually Temperature Independent. Biophys J 2020; 119:2584-2592. [PMID: 33189678 DOI: 10.1016/j.bpj.2020.10.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/06/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
The voltage-dependent anion channel (VDAC) is the most abundant protein in the mitochondrial outer membrane and an archetypical β-barrel channel. Here, we study the effects of temperature on VDAC channels reconstituted in planar lipid membranes at the single- and multichannel levels within the 20°C to 40°C range. The temperature dependence of conductance measured on a single channel in 1 M KCl shows an increase characterized by a 10°C temperature coefficient Q10 = 1.22 ± 0.02, which exceeds that of the bathing electrolyte solution conductivity, Q10 = 1.17 ± 0.01. The rates of voltage-induced channel transition between the open and closed states measured on multichannel membranes also show statistically significant increases, with temperatures that are consistent with activation energy barriers of ∼10 ± 3 kcal/mol. At the same time, the gating thermodynamics, as characterized by the gating charge and voltage of equipartitioning, does not display any measurable temperature dependence. The two parameters stay within 3.2 ± 0.2 elementary charges and 30 ± 2 mV, respectively. Thus, whereas the channel kinetics, specifically its conductance and rates of gating response to voltage steps, demonstrates a clear increase with temperature, the conformational voltage-dependent equilibria are virtually insensitive to temperature. These results, which may be a general feature of β-barrel channel gating, suggest either an entropy-driven gating mechanism or a role for enthalpy-entropy compensation.
Collapse
|
47
|
Xiao R, Xu XZS. Temperature Sensation: From Molecular Thermosensors to Neural Circuits and Coding Principles. Annu Rev Physiol 2020; 83:205-230. [PMID: 33085927 DOI: 10.1146/annurev-physiol-031220-095215] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Temperature is a universal cue and regulates many essential processes ranging from enzymatic reactions to species migration. Due to the profound impact of temperature on physiology and behavior, animals and humans have evolved sophisticated mechanisms to detect temperature changes. Studies from animal models, such as mouse, Drosophila, and C. elegans, have revealed many exciting principles of thermosensation. For example, conserved molecular thermosensors, including thermosensitive channels and receptors, act as the initial detectors of temperature changes across taxa. Additionally, thermosensory neurons and circuits in different species appear to adopt similar logic to transduce and process temperature information. Here, we present the current understanding of thermosensation at the molecular and cellular levels. We also discuss the fundamental coding strategies of thermosensation at the circuit level. A thorough understanding of thermosensation not only provides key insights into sensory biology but also builds a foundation for developing better treatments for various sensory disorders.
Collapse
Affiliation(s)
- Rui Xiao
- Department of Aging and Geriatric Research, Institute on Aging and Center for Smell and Taste, University of Florida, Gainesville, Florida 32610, USA;
| | - X Z Shawn Xu
- Life Sciences Institute and Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA;
| |
Collapse
|
48
|
Mukherjee S, Sikdar SK. Intracellular activation of full-length human TREK-1 channel by hypoxia, high lactate, and low pH denotes polymodal integration by ischemic factors. Pflugers Arch 2020; 473:167-183. [PMID: 33025137 DOI: 10.1007/s00424-020-02471-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/18/2020] [Accepted: 09/30/2020] [Indexed: 10/23/2022]
Abstract
TREK-1, a two-pore domain potassium channel, responds to ischemic levels of intracellular lactate and acidic pH to provide neuroprotection. There are two splice variants of hTREK1: the shorter splice variant having a shorter N-terminus compared with the full-length hTREK1 with similar C-terminus sequence that is widely expressed in the brain. The shorter variant was reported to be irresponsive to hypoxia-a condition attributed to ischemia, which has put the neuroprotective role of hTREK-1 channel into question. Since interaction between N- and C-terminus of different ion channels shapes their gating, we re-examined the sensitivity of the full-length as well as the shorter hTREK-1 channel to intracellular hypoxia along with lactate. Single-channel data obtained from the excised inside-out patches of the full-length channel expressed in HEK293 cells indicated an increase in activity as opposed to a decrease in activity in the shorter isoform. However, both the isoforms showed an increase in activity under combined hypoxia, 20mM lactate, and low pH 6 condition, albeit with subtle differences in their individual actions, confirming the neuroprotective role played by hTREK-1 irrespective of the differences in the N-terminus among the splice variants. Furthermore, E321A mutant that disrupts the interaction of the C-terminus with the membrane showed a decrease in activity with hypoxia indicating the importance of the C-terminus in the hypoxic response of the full-length hTREK-1. We propose an increase in activity of both the splice variants of hTREK-1 in combined hypoxia, high lactate, and low pH conditions typically associated with ischemia provides neuroprotection.
Collapse
Affiliation(s)
- Sourajit Mukherjee
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Sujit Kumar Sikdar
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, 560012, India.
| |
Collapse
|
49
|
Buijs TJ, McNaughton PA. The Role of Cold-Sensitive Ion Channels in Peripheral Thermosensation. Front Cell Neurosci 2020; 14:262. [PMID: 32973456 PMCID: PMC7468449 DOI: 10.3389/fncel.2020.00262] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/27/2020] [Indexed: 11/13/2022] Open
Abstract
The detection of ambient cold is critical for mammals, who use this information to avoid tissue damage by cold and to maintain stable body temperature. The transduction of information about the environmental cold is mediated by cold-sensitive ion channels expressed in peripheral sensory nerve endings in the skin. Most transduction mechanisms for detecting temperature changes identified to date depend on transient receptor potential (TRP) ion channels. Mild cooling is detected by the menthol-sensitive TRPM8 ion channel, but how painful cold is detected remains unclear. The TRPA1 ion channel, which is activated by cold in expression systems, seemed to provide an answer to this question, but whether TRPA1 is activated by cold in neurons and contributes to the sensation of cold pain continues to be a matter of debate. Recent advances have been made in this area of investigation with the identification of several potential cold-sensitive ion channels in thermosensory neurons, including two-pore domain potassium channels (K2P), GluK2 glutamate receptors, and CNGA3 cyclic nucleotide-gated ion channels. This mini-review gives a brief overview of the way by which ion channels contribute to cold sensation, discusses the controversy around the cold-sensitivity of TRPA1, and provides an assessment of some recently-proposed novel cold-transduction mechanisms. Evidence for another unidentified cold-transduction mechanism is also presented.
Collapse
Affiliation(s)
- Tamara Joëlle Buijs
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | | |
Collapse
|
50
|
A folding reaction at the C-terminal domain drives temperature sensing in TRPM8 channels. Proc Natl Acad Sci U S A 2020; 117:20298-20304. [PMID: 32747539 DOI: 10.1073/pnas.2004303117] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In mammals, temperature-sensitive TRP channels make membrane conductance of cells extremely temperature dependent, allowing the detection of temperature ranging from noxious cold to noxious heat. We progressively deleted the distal carboxyl terminus domain (CTD) of the cold-activated melastatin receptor channel, TRPM8. We found that the enthalpy change associated with channel gating is proportional to the length of the CTD. Deletion of the last 36 amino acids of the CTD transforms TRPM8 into a reduced temperature-sensitivity channel (Q10 ∼4). Exposing the intracellular domain to a denaturing agent increases the energy required to open the channel indicating that cold drives channel gating by stabilizing the folded state of the CTD. Experiments in the presence of an osmoticant agent suggest that channel gating involves a change in solute-inaccessible volume in the CTD of ∼1,900 Å3 This volume matches the void space inside the coiled coil according to the cryogenic electron microscopy structure of TRPM8. The results indicate that a folding-unfolding reaction of a specialized temperature-sensitive structure is coupled to TRPM8 gating.
Collapse
|